Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Sci Rep ; 13(1): 19538, 2023 11 09.
Artigo em Inglês | MEDLINE | ID: mdl-37945645

RESUMO

Neonatal hyperoxia induces long-term systemic vascular stiffness and cardiovascular remodeling, but the mechanisms are unclear. Chemokine receptor 7 (CXCR7) represents a key regulator of vascular homeostasis and repair by modulating TGF-ß1 signaling. This study investigated whether pharmacological CXCR7 agonism prevents neonatal hyperoxia-induced systemic vascular stiffness and cardiac dysfunction in juvenile rats. Newborn Sprague Dawley rat pups assigned to room air or hyperoxia (85% oxygen), received CXCR7 agonist, TC14012 or placebo for 3 weeks. These rat pups were maintained in room air until 6 weeks when aortic pulse wave velocity doppler, cardiac echocardiography, aortic and left ventricular (LV) fibrosis were assessed. Neonatal hyperoxia induced systemic vascular stiffness and cardiac dysfunction in 6-week-old rats. This was associated with decreased aortic and LV CXCR7 expression. Early treatment with TC14012, partially protected against neonatal hyperoxia-induced systemic vascular stiffness and improved LV dysfunction and fibrosis in juvenile rats by decreasing TGF-ß1 expression. In vitro, hyperoxia-exposed human umbilical arterial endothelial cells and coronary artery endothelial cells had increased TGF-ß1 levels. However, treatment with TC14012 significantly reduced the TGF-ß1 levels. These results suggest that dysregulation of endothelial CXCR7 signaling may contribute to neonatal hyperoxia-induced systemic vascular stiffness and cardiac dysfunction.


Assuntos
Hiperóxia , Disfunção Ventricular Esquerda , Animais , Humanos , Ratos , Animais Recém-Nascidos , Células Endoteliais , Fibrose , Hiperóxia/complicações , Análise de Onda de Pulso , Ratos Sprague-Dawley , Fator de Crescimento Transformador beta1 , Remodelação Vascular
2.
Front Cell Dev Biol ; 11: 1245747, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-38481391

RESUMO

Background: Intra-amniotic inflammation (IAI) is associated with increased risk of preterm birth and bronchopulmonary dysplasia (BPD), but the mechanisms by which IAI leads to preterm birth and BPD are poorly understood, and there are no effective therapies for preterm birth and BPD. The transcription factor c-Myc regulates various biological processes like cell growth, apoptosis, and inflammation. We hypothesized that c-Myc modulates inflammation at the maternal-fetal interface, and neonatal lung remodeling. The objectives of our study were 1) to determine the kinetics of c-Myc in the placenta, fetal membranes and neonatal lungs exposed to IAI, and 2) to determine the role of c-Myc in modulating inflammation at the maternal-fetal interface, and neonatal lung remodeling induced by IAI. Methods: Pregnant Sprague-Dawley rats were randomized into three groups: 1) Intra-amniotic saline injections only (control), 2) Intra-amniotic lipopolysaccharide (LPS) injections only, and 3) Intra-amniotic LPS injections with c-Myc inhibitor 10058-F4. c-Myc expression, markers of inflammation, angiogenesis, immunohistochemistry, and transcriptomic analyses were performed on placenta and fetal membranes, and neonatal lungs to determine kinetics of c-Myc expression in response to IAI, and effects of prenatal systemic c-Myc inhibition on lung remodeling at postnatal day 14. Results: c-Myc was upregulated in the placenta, fetal membranes, and neonatal lungs exposed to IAI. IAI caused neutrophil infiltration and neutrophil extracellular trap (NET) formation in the placenta and fetal membranes, and neonatal lung remodeling with pulmonary hypertension consistent with a BPD phenotype. Prenatal inhibition of c-Myc with 10058-F4 in IAI decreased neutrophil infiltration and NET formation, and improved neonatal lung remodeling induced by LPS, with improved alveolarization, increased angiogenesis, and decreased pulmonary vascular remodeling. Discussion: In a rat model of IAI, c-Myc regulates neutrophil recruitment and NET formation in the placenta and fetal membranes. c-Myc also participates in neonatal lung remodeling induced by IAI. Further studies are needed to investigate c-Myc as a potential therapeutic target for IAI and IAI-associated BPD.

3.
Lung ; 200(4): 463-472, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35717488

RESUMO

PURPOSE: Alpha-melanocyte stimulating hormone (α-MSH) is known to have anti-inflammatory effects. However, the anti-inflammatory properties of α-MSH on normal bronchial epithelial cells are largely unknown, especially in the context of in vitro sarcoidosis models. METHODS: We evaluated the anti-inflammatory effects of α-MSH on two different in vitro sarcoidosis models (lung-on-membrane model; LOMM and three-dimensional biochip pulmonary sarcoidosis model; 3D-BSGM) generated from NBECs and an in vivo sarcoidosis mouse model. RESULTS: Treatment with α-MSH decreased inflammatory cytokine levels and downregulated type I interferon pathway genes and related proteins in LOMM and 3D-BSGM models. Treatment with α-MSH also significantly decreased macrophages and cytotoxic T-cells counts in a sarcoidosis mice model. CONCLUSION: Our results confirm the direct role of type I IFNs in the pathogenesis of sarcoid lung granulomas and highlight α-MSH as a potential novel therapeutic agent for treating pulmonary sarcoidosis.


Assuntos
Sarcoidose Pulmonar , Sarcoidose , Animais , Anti-Inflamatórios/farmacologia , Anti-Inflamatórios/uso terapêutico , Granuloma/tratamento farmacológico , Inflamação/metabolismo , Camundongos , Sarcoidose/tratamento farmacológico , Sarcoidose Pulmonar/tratamento farmacológico , alfa-MSH/metabolismo , alfa-MSH/farmacologia , alfa-MSH/uso terapêutico
4.
Clin Transl Immunology ; 10(7): e1310, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34257968

RESUMO

OBJECTIVES: Growth hormone-releasing hormone (GHRH) is a potent stimulator of growth hormone (GH) secretion from the pituitary gland. Although GHRH is essential for the growth of immune cells, the regulatory effects of its antagonist in granulomatous disease remain unknown. METHODS: Here, we report expression of GHRH receptor (R) in human tissue with sarcoidosis granuloma and demonstrate the anti-inflammatory effects of MIA602 (a GHRH antagonist) in two in vitro human granuloma models and an in vivo granuloma model using different methods including ELISA, immunohistochemistry, RNA-seq analysis and flow cytometry. RESULTS: MIA602 decreases the levels of IL-2, IL-2R, IL-7, IL-12, IL-17A and TNF-α in an in vitro granuloma model. Further, we show that the anti-inflammatory effect of MIA602 appears to be mediated by a reduction in CD45+CD68+ cells in granulomatous tissue and upregulation in PD-1 expression in macrophages. Analysis of the expression of proteins involved in the mitochondrial stage of apoptosis showed that MIA602 increases the levels of caspase-3, BCL-xL/BAK dimer and MCl-1/Bak dimer in the granuloma. These findings indicate that MIA602 may not induce apoptosis. CONCLUSIONS: Our findings further suggest that GHRH-R is potentially a clinical target for the treatment of granulomatous disease and that MIA602 may be used as a novel therapeutic agent for sarcoidosis.

5.
PLoS One ; 16(2): e0245805, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33539409

RESUMO

Sarcoidosis is a multi-system disorder of granulomatous inflammation which most commonly affects the lungs. Its etiology and pathogenesis are not well defined in part due to the lack of reliable modeling. Here, we present the development of an in vitro three-dimensional lung-on-chip biochip designed to mimic granuloma formation. A lung on chip fluidic macrodevice was developed and added to our previously developed a lung-on-membrane model (LOMM). Granulomas were cultured from blood samples of patients with sarcoidosis and then inserted in the air-lung-interface of the microchip to create a three-dimensional biochip pulmonary sarcoidosis model (3D BSGM). Cytokines were measured after 48 hours. ELISA testing was performed to measure cytokine response difference between LOMM with 3D BSGM. There were statistically significant differences in IL-1ß (P = 0.001953), IL-6 (P = 0.001953), GM-CSF (P = 0.001953), and INF-γ expressions (P = 0.09375) between two groups. The current model represents the first 3D biochip sarcoidosis model created by adding a microfluidics system to a dual-chambered lung on membrane model and introducing developed sarcoid-granuloma to its air-lung-interface.


Assuntos
Biomimética/instrumentação , Dispositivos Lab-On-A-Chip , Sarcoidose Pulmonar/patologia , Citocinas/metabolismo , Humanos , Leucócitos Mononucleares/metabolismo
6.
Sci Rep ; 11(1): 3455, 2021 02 10.
Artigo em Inglês | MEDLINE | ID: mdl-33568776

RESUMO

The Nucleocapsid Protein (N Protein) of severe acute respiratory syndrome Coronavirus 2 (SARS-CoV2) is located in the viral core. Immunoglobulin G (IgG) targeting N protein is detectable in the serum of infected patients. The effect of high titers of IgG against N-protein on clinical outcomes of SARS-CoV2 disease has not been described. We studied 400 RT-PCR confirmed SARS-CoV2 patients to determine independent factors associated with poor outcomes, including Medical Intensive Care Unit (MICU) admission, prolonged MICU stay and hospital admissions, and in-hospital mortality. We also measured serum IgG against the N protein and correlated its concentrations with clinical outcomes. We found that several factors, including Charlson comorbidity Index (CCI), high levels of IL6, and presentation with dyspnea were associated with poor clinical outcomes. It was shown that higher CCI and higher IL6 levels were independently associated with in-hospital mortality. Anti-N protein IgG was detected in the serum of 55 (55%) patients at the time of admission. A high concentration of antibodies, defined as signal to cut off ratio (S/Co) > 1.5 (75 percentile of all measurements), was found in 25 (25%) patients. The multivariable logistic regression models showed that between being an African American, higher CCI, lymphocyte counts, and S/Co ratio > 1.5, only S/Co ratio were independently associated with MICU admission and longer length of stay in hospital. This study recommends that titers of IgG targeting N-protein of SARS-CoV2 at admission is a prognostic factor for the clinical course of disease and should be measured in all patients with SARS-CoV2 infection.


Assuntos
COVID-19/imunologia , Imunoglobulina G/imunologia , Proteínas do Nucleocapsídeo/imunologia , SARS-CoV-2/imunologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Anticorpos Antivirais/sangue , Anticorpos Antivirais/imunologia , COVID-19/sangue , Feminino , Humanos , Imunoglobulina G/sangue , Unidades de Terapia Intensiva , Tempo de Internação , Masculino , Pessoa de Meia-Idade , Prognóstico , Glicoproteína da Espícula de Coronavírus/imunologia
7.
Am J Physiol Renal Physiol ; 320(3): F442-F453, 2021 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-33459165

RESUMO

Diabetic nephropathy (DN) is the leading cause of end-stage kidney disease. Besides glycemic and blood pressure control, environmental factors such as cigarette smoking (CS) adversely affect the progression of DN. The effects of CS on DN progression have been attributed to combustion-generated molecules without consideration to the role of nicotine (NIC), responsible for the addictive properties of both CS and electronic cigarettes (ECs). Podocytes are essential to preserve the structure and function of the glomerular filtration barrier, and strong evidence indicates that early podocyte loss promotes DN progression. We performed experiments in human podocytes and in a mouse model of diabetes that develops nephropathy resembling human DN. We determined that NIC binding to podocytes in concentrations achieved with CS and ECs activated NADPH oxidase, which sets in motion a dysfunctional molecular network integrated by cyclooxygenase 2, known to induce podocyte injury; downregulation of AMP-activated protein kinase, important for maintaining cellular energy stores and antioxidation; and upregulation of CD36, which increased lipid uptake and promoted apoptosis. In diabetic mice, NIC increased proteinuria, a recognized marker of chronic kidney disease progression, accompanied by reduced glomerular podocyte synaptopodin, a crucial stabilizer of the podocyte cytoskeleton, and increased fibronectin expression. This novel study critically implicates NIC itself as a contributor to DN progression in CS and EC users.NEW & NOTEWORTHY In this study, we demonstrate that nicotine increases the production of reactive oxygen species, increases cyclooxygenase-2 expression, and upregulates Cd36 while inducing downregulation of AMP-activated protein kinase. In vivo nicotine increases proteinuria and fibronectin expression in diabetic mice. This study demonstrates that effects of nicotine on podocytes are responsible, at least in part, for the deleterious effects of smoking in the progression of chronic kidney disease, including diabetic nephropathy.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Nefropatias Diabéticas/metabolismo , Nicotina/farmacologia , Podócitos/metabolismo , Fumar/efeitos adversos , Animais , Apoptose/efeitos dos fármacos , Diabetes Mellitus Experimental/metabolismo , Nefropatias Diabéticas/tratamento farmacológico , Humanos , Camundongos , Podócitos/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo
8.
J Vasc Surg ; 68(6S): 127S-135S, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30064832

RESUMO

OBJECTIVE: Endothelial progenitor cells (EPCs) are the key cells of postnatal neovascularization, and mesenchymal stem cells (MSCs) possess pluripotent differentiation capacity and contribute to tissue regeneration and wound healing. Both EPCs and MSCs are critical to the wound repair process, which is hindered in diabetes mellitus. Diabetes has been shown to decrease the function of these progenitor cells, whereas estrogen has beneficial wound healing effects. However, the role of estrogen in modulating EPC and MSC biology in diabetes is unknown. We investigated the effect of estrogen on improving bone marrow (BM)-derived EPC and MSC function using a murine diabetic wound healing model. METHODS: Female diabetic db+/db+ and nondiabetic control mice were wounded cutaneously and treated with topical estrogen or placebo cream. On day 5 after wounding, BM cells were harvested to quantify EPC number and colony-forming units of EPCs and MSCs. Wound healing rate was concurrently studied. Vessel density and scar density were then quantified using whole body perfusion and laser confocal microscopy. EPC recruitment was documented by immunohistochemistry to identify CD34- and vascular endothelial growth factor receptor 2-positive cells in the vessel wall. Data were analyzed by analysis of variance. RESULTS: Topical estrogen significantly increased colony-forming units of both EPCs and MSCs compared with placebo treatment, indicating improved viability and proliferative ability of these cells. Consistently, increased recruitment of EPCs to diabetic wounds and higher vessel density were observed in estrogen-treated compared with placebo-treated mice. Consequently, topical estrogen significantly accelerated wound healing as early as day 6 after wounding. In addition, scar density resulting from collagen deposition was increased in the estrogen-treated group, reflecting increased MSC activity and differentiation. CONCLUSIONS: Estrogen treatment increases wound healing and wound neovascularization in diabetic mice. Our data implicate that these beneficial effects may be mediated through improving the function of BM-derived EPCs and MSCs.


Assuntos
Diabetes Mellitus , Células Progenitoras Endoteliais/efeitos dos fármacos , Estrogênios/administração & dosagem , Células-Tronco Mesenquimais/efeitos dos fármacos , Pele/irrigação sanguínea , Pele/efeitos dos fármacos , Cicatrização/efeitos dos fármacos , Ferimentos Penetrantes/tratamento farmacológico , Administração Cutânea , Animais , Antígenos CD34/metabolismo , Proliferação de Células/efeitos dos fármacos , Colágeno/metabolismo , Diabetes Mellitus/metabolismo , Diabetes Mellitus/patologia , Células Progenitoras Endoteliais/metabolismo , Células Progenitoras Endoteliais/patologia , Feminino , Células-Tronco Mesenquimais/metabolismo , Células-Tronco Mesenquimais/patologia , Camundongos Mutantes , Neovascularização Fisiológica/efeitos dos fármacos , Pomadas , Fenótipo , Pele/lesões , Pele/metabolismo , Fatores de Tempo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Ferimentos Penetrantes/metabolismo , Ferimentos Penetrantes/patologia
9.
PLoS One ; 12(12): e0188439, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29236702

RESUMO

Obesity and cigarette smoke are major cardiovascular (CV) risk factors and, when coexisting in the same individuals, have additive/synergistic effects upon CVD. We studied the mechanisms involved in nicotine enhancement of CVD in Sprague Dawley rats with diet-induced obesity. The rats were fed either a high fat (HFD) or normal rat chow diet with or without nicotine (100 mg/L in drinking water) for 20 weeks. HFD rats developed central obesity, increased systolic blood pressure (SBP), aortic superoxide (O2-) production, and impaired endothelial nitric oxide synthase (eNOS) and endothelium-dependent relaxation to acetylcholine (EDR). Nicotine further increased SBP, O2- and impaired eNOS and EDR in obese rats. In the peritoneal macrophages from obese rats, tumor necrosis factor (TNF) α, interleukin 1ß and CD36 were increased, and were further increased in nicotine-treated obese rats. Using PCR array we found that 3 of 84 target proinflammatory genes were increased by 2-4 fold in the aorta of obese rats, 11 of the target genes were further increased in nicotine-treated obese rats. HUVECs, incubated with conditioned medium from the peritoneal macrophages of nicotine treated-obese rats, exhibited reduced eNOS and increased NADPH oxidase subunits gp91phox and p22phox expression. Those effects were partially prevented by adding anti-TNFα antibody to the conditioned medium. Our results suggest that nicotine aggravates the CV effects of diet-induced obesity including the oxidative stress, vascular inflammation and endothelial dysfunction. The underlying mechanisms may involve in targeting endothelium by enhancement of macrophage-derived TNFα.


Assuntos
Cotinina/sangue , Dieta Hiperlipídica/efeitos adversos , Endotélio Vascular/efeitos dos fármacos , Macrófagos Peritoneais/metabolismo , Nicotina/farmacologia , Obesidade/fisiopatologia , Fator de Necrose Tumoral alfa/fisiologia , Vasculite/fisiopatologia , Administração Oral , Animais , Pressão Sanguínea/efeitos dos fármacos , Peso Corporal/efeitos dos fármacos , Proteína C-Reativa/metabolismo , Endotélio Vascular/enzimologia , Endotélio Vascular/metabolismo , Endotélio Vascular/fisiopatologia , Masculino , Nicotina/administração & dosagem , Obesidade/complicações , Obesidade/etiologia , Ratos , Ratos Sprague-Dawley , Espécies Reativas de Oxigênio/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Vasculite/complicações
10.
Sci Rep ; 6: 34416, 2016 10 07.
Artigo em Inglês | MEDLINE | ID: mdl-27713493

RESUMO

Homing of endothelial progenitor cells (EPC) to the ischemic tissues is a key event in neovascularization and tissue regeneration. In response to ischemic insult, injured tissues secrete several chemo-cytokines, including stromal cell-derived factor-1α (SDF-1α), which triggers mobilization and homing of bone marrow-derived EPC (BMD-EPC). We previously reported that SDF-1α-induced EPC homing is mediated by a panel of adhesion molecules highly or selectively expressed on the activated endothelium in ischemic tissues, including E-selectin. Elevated E-selectin on wound vasculature serve as docking sites for circulating EPC, which express counterpart E-selectin ligands. Here, we show that SDF-1α presented in wound tissue and released into circulation can act both locally and remotely to induce ischemic tissue endothelium and BMD-EPC to express both E-selectin and its ligands. By performing BM transplantation using E-selectin-/- and E-selectin+/+ mice as the donors and recipients respectively, we demonstrate that upregulated dual E-selectin/ligand pairs reciprocally expressed on ischemic tissue endothelium and BMD-EPC act as double-locks to secure targeted EPC- endothelium interactions by which to facilitate EPC homing and promote neovascularization and tissue repair. These findings describe a novel mechanism for BMD-EPC homing and indicate that dual E-selectin/ligand pairs may be effective targets/tools for therapeutic neovascularization and targeted cell delivery.


Assuntos
Quimiocina CXCL12/metabolismo , Selectina E/metabolismo , Células Progenitoras Endoteliais/metabolismo , Isquemia/metabolismo , Neovascularização Patológica , Cicatrização , Animais , Adesão Celular , Movimento Celular , Células Cultivadas , Quimiocina CXCL12/administração & dosagem , Selectina E/genética , Humanos , Receptores de Hialuronatos/metabolismo , Camundongos , Camundongos Knockout , Receptores de Fatores de Crescimento de Fibroblastos , Sialoglicoproteínas
11.
Am J Physiol Renal Physiol ; 311(6): F1308-F1317, 2016 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-27335373

RESUMO

Podocytes are the key target for injury in proteinuric glomerular diseases that result in podocyte loss, progressive focal segmental glomerular sclerosis (FSGS), and renal failure. Current evidence suggests that the initiation of podocyte injury and associated proteinuria can be separated from factors that drive and maintain these pathogenic processes leading to FSGS. In nephrotic urine aberrant glomerular filtration of plasminogen (Plg) is activated to the biologically active serine protease plasmin by urokinase-type plasminogen activator (uPA). In vivo inhibition of uPA mitigates Plg activation and development of FSGS in several proteinuric models of renal disease including 5/6 nephrectomy. Here, we show that Plg is markedly increased in the urine in two murine models of proteinuric kidney disease associated with podocyte injury: Tg26 HIV-associated nephropathy and the Cd2ap-/- model of FSGS. We show that human podocytes express uPA and three Plg receptors: uPAR, tPA, and Plg-RKT. We demonstrate that Plg treatment of podocytes specifically upregulates NADPH oxidase isoforms NOX2/NOX4 and increases production of mitochondrial-dependent superoxide anion (O2-) that promotes endothelin-1 synthesis. Plg via O2- also promotes expression of the B scavenger receptor CD36 and subsequent increased intracellular cholesterol uptake resulting in podocyte apoptosis. Taken together, our findings suggest that following disruption of the glomerular filtration barrier at the onset of proteinuric disease, podocytes are exposed to Plg resulting in further injury mediated by oxidative stress. We suggest that chronic exposure to Plg could serve as a "second hit" in glomerular disease and that Plg is potentially an attractive target for therapeutic intervention.


Assuntos
Rim/metabolismo , Estresse Oxidativo/fisiologia , Plasminogênio/urina , Podócitos/metabolismo , Proteinúria/metabolismo , Nefropatia Associada a AIDS/metabolismo , Nefropatia Associada a AIDS/patologia , Animais , Modelos Animais de Doenças , Glomerulosclerose Segmentar e Focal/metabolismo , Glomerulosclerose Segmentar e Focal/patologia , Rim/patologia , Falência Renal Crônica/metabolismo , Falência Renal Crônica/patologia , Camundongos , NADPH Oxidases/metabolismo , Podócitos/patologia , Proteinúria/patologia , Regulação para Cima , Ativador de Plasminogênio Tipo Uroquinase/metabolismo
12.
PLoS One ; 11(4): e0154053, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27104647

RESUMO

Stem cell therapy has emerged as a promising approach for treatment of a number of diseases, including delayed and non-healing wounds. However, targeted systemic delivery of therapeutic cells to the dysfunctional tissues remains one formidable challenge. Herein, we present a targeted nanocarrier-mediated cell delivery method by coating the surface of the cell to be delivered with dendrimer nanocarriers modified with adhesion molecules. Infused nanocarrier-coated cells reach to destination via recognition and association with the counterpart adhesion molecules highly or selectively expressed on the activated endothelium in diseased tissues. Once anchored on the activated endothelium, nanocarriers-coated transporting cells undergo transendothelial migration, extravasation and homing to the targeted tissues to execute their therapeutic role. We now demonstrate feasibility, efficacy and safety of our targeted nanocarrier for delivery of bone marrow cells (BMC) to cutaneous wound tissues and grafted corneas and its advantages over conventional BMC transplantation in mouse models for wound healing and neovascularization. This versatile platform is suited for targeted systemic delivery of virtually any type of therapeutic cell.


Assuntos
Selectina E/metabolismo , Neovascularização Fisiológica , Células-Tronco/citologia , Cicatrização , Animais , Membrana Celular/metabolismo , Dendrímeros , Células Endoteliais da Veia Umbilical Humana , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Microscopia Eletrônica de Transmissão
13.
Cardiovasc Diabetol ; 14: 45, 2015 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-25928697

RESUMO

BACKGROUND: We have previously shown that in hypertensive Dahl salt-sensitive (DS) rats, impaired endothelium-dependent relaxation to acetylcholine and to insulin is mechanistically linked to up-regulation of angiotensin (Ang) II actions and the production of reactive oxygen species (ROS) and to activation of the proinflammatory transcription factor (NF)κB. Here we investigated whether Ang II activation of NFκB contributed to insulin resistance in the skeletal muscle of this animal model. METHODS: DS rats were fed either a normal (NS, 0.5% NaCl) or high (HS, 4% NaCl) salt diet for 6 weeks. In addition, 3 separate groups of HS rats were given angiotensin receptor 1 blocker candesartan (ARB, 10 mg/kg/day in drinking water), antioxidant tempol (1 mmol/L in drinking water) or NFκB inhibitor PDTC (150 mg/kg in drinking water). RESULTS: DS rats manifested an increase in soleus muscle Ang II content, ROS production and phosopho-IκBα/IκBα ratio, ARB or tempol reduced ROS and phospho-IκBα/IκBα ratio. Hypertensive DS rats also manifested a reduction in glucose infusion rate, impaired insulin-induced Akt phosphorylation and Glut-4 translocation in the soleus muscle, which were prevented with treatment of either ARB, tempol, or PDTC. Data from the rat diabetes signaling pathway PCR array showed that 8 genes among 84 target genes were altered in the muscle of hypertensive rats with the increase in gene expression of ACE1 and 5 proinflammatory genes, and decrease of 2 glucose metabolic genes. Incubation of the muscle with NFκB SN50 (a specific peptide inhibitor of NFκB) ex vivo reversed changes in hypertension-induced gene expression. CONCLUSION: The current findings strongly suggest that the activation of NFκB inflammatory pathway by Ang II play a critical role in skeletal muscle insulin resistance in salt-sensitive hypertension.


Assuntos
Angiotensina II/metabolismo , Hipertensão/metabolismo , Resistência à Insulina/fisiologia , Músculo Esquelético/metabolismo , NF-kappa B/metabolismo , Cloreto de Sódio na Dieta/toxicidade , Animais , Hipertensão/induzido quimicamente , Masculino , Ratos , Ratos Endogâmicos Dahl , Ratos Sprague-Dawley
14.
Am J Hypertens ; 27(6): 873-80, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24413709

RESUMO

BACKGROUND: Current treatment for the secondary prevention of cardiovascular diseases frequently involves the prescription of several combination therapies, particularly antihypertensive medications and HMG-CoA reductase inhibitor. We have previously shown that in salt-sensitive hypertension either a statin or the calcium channel blocker amlodipine (Aml) have vasoprotective effects. Here, we investigated in aortas from Dahl salt-sensitive (DS) rats the effects of Aml, the statin atorvastatin (AT), and their combination on endothelial function, superoxide (O2 (-)) production, and the expression of endothelial nitric oxide synthase (eNOS), chemokine monocyte chemoattractant protein-1 (MCP-1), and lectin-like oxidized LDL receptor-1 (LOX-1). METHODS: Groups of DS rats were fed either normal-salt (NS, 0.5% NaCl) or high-salt (HS, 4% NaCl) diet or a HS diet with AT (15mg/kg/day), Aml (5mg/kg/day) or combination of AT/Aml for 6 weeks. RESULTS: Rats on the HS diet developed hypertension, aortic hypertrophy, accompanied by increased plasma C-reactive protein (CRP), aortic O2 (-), MCP-1 (80%), and LOX-1 (55%) expression and reduced eNOS and endothelial-dependent relaxation to acetylcholine (EDR). Aml reduced systolic blood pressure (SBP), aortic hypertrophy, plasma CRP, vascular O2 (-), and MCP-1 expression and improved eNOS and EDR. AT reduced aortic hypertrophy and plasma CRP, improved EDR, and normalized vascular O2 (-), eNOS, and proinflammatory gene expression with mild reduction in SBP. Combination therapy further reduced the SBP and normalized aortic hypertrophy, EDR, and plasma CRP. CONCLUSIONS: The combination therapy of Aml/AT has an additive beneficial effect on the vasculature. These novel findings may provide scientific basis for the combination therapy of statins with antihypertensive agents to reduce and prevent cardiovascular diseases.


Assuntos
Anlodipino/farmacologia , Anti-Hipertensivos/farmacologia , Aorta/efeitos dos fármacos , Bloqueadores dos Canais de Cálcio/farmacologia , Ácidos Heptanoicos/farmacologia , Inibidores de Hidroximetilglutaril-CoA Redutases/farmacologia , Hipertensão/tratamento farmacológico , Pirróis/farmacologia , Cloreto de Sódio na Dieta , Animais , Aorta/metabolismo , Aorta/patologia , Aorta/fisiopatologia , Atorvastatina , Pressão Sanguínea/efeitos dos fármacos , Proteína C-Reativa/metabolismo , Quimiocina CCL2/metabolismo , Modelos Animais de Doenças , Quimioterapia Combinada , Hipertensão/etiologia , Hipertensão/metabolismo , Hipertensão/patologia , Hipertensão/fisiopatologia , Hipertrofia , Masculino , Óxido Nítrico Sintase Tipo III/metabolismo , Ratos Endogâmicos Dahl , Receptores Depuradores Classe E/metabolismo , Superóxidos/metabolismo , Fatores de Tempo , Vasodilatação/efeitos dos fármacos
15.
Atherosclerosis ; 225(2): 296-303, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23078884

RESUMO

OBJECTIVE: Notch signaling plays pivotal roles in the pathogenesis of vascular disease. However, little is known about its role in atherosclerosis. We sought to investigate the potential involvement of the Notch signaling in atherosclerosis. METHODS: Expression of Notch pathway components in mouse and human aorta with or without atherosclerosis plaque was examined by immunohistochemistry. Expression of Notch target genes in young versus aged human endothelial cells (EC) was examined by PCRArray and immunoblot. In vitro loss- and gain-of-function approaches were utilized to evaluate the role of Notch signaling in inducing EC senescence and secretion of pro-inflammatory cytokines by ProteinArray. Notch gene profile was studied in 1054 blood samples of patients with coronary artery disease (CAD). Genotyping was performed using the Genome-Wide Single Nucleotide Polymorphism (SNP) Array. RESULTS: Notch pathway components were upregulated in luminal EC at atherosclerotic lesions from mouse and human aortas. In addition, the Notch pathway was activated in aged but not young human EC. Enforced Notch activation resulted in EC senescence and significantly upregulated expression of several molecules implicated in the inflammatory response (IL-6/IL-8/IL-1α/RANTES/ICAM-1). The upregulated IL-6 was partially responsible for mediating leukocyte transendothelial migration. Genetic association analysis detected, of 82 SNPs across 6 Notch pathway genes analyzed, 4 SNPs with nominal association with CAD burden. CONCLUSION: Notch pathway is activated in luminal EC at atherosclerotic plaques and results in pro-inflammatory response and senescence of EC. Notch signaling may be linked to human CAD risk. These findings implicate a potential involvement of Notch signaling in atherosclerosis.


Assuntos
Aterosclerose/metabolismo , Senescência Celular , Citocinas/metabolismo , Células Endoteliais/metabolismo , Mediadores da Inflamação/metabolismo , Inflamação/metabolismo , Receptores Notch/metabolismo , Transdução de Sinais , Fatores Etários , Animais , Aorta/metabolismo , Aorta/patologia , Doenças da Aorta/genética , Doenças da Aorta/metabolismo , Doenças da Aorta/patologia , Apolipoproteínas E/deficiência , Apolipoproteínas E/genética , Aterosclerose/genética , Aterosclerose/patologia , Western Blotting , Doença da Artéria Coronariana/genética , Doença da Artéria Coronariana/metabolismo , Doença da Artéria Coronariana/patologia , Vasos Coronários/metabolismo , Vasos Coronários/patologia , Modelos Animais de Doenças , Células Endoteliais/patologia , Perfilação da Expressão Gênica , Células HEK293 , Humanos , Inflamação/genética , Inflamação/patologia , Leucócitos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Células NIH 3T3 , Placa Aterosclerótica , Reação em Cadeia da Polimerase , Polimorfismo de Nucleotídeo Único , Análise Serial de Proteínas , Receptores Notch/genética , Transdução de Sinais/genética , Fatores de Tempo , Migração Transendotelial e Transepitelial , Transfecção , Regulação para Cima
16.
Ann Surg ; 256(4): 560-72, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22964729

RESUMO

OBJECTIVES: We have previously shown that stromal cell-derived factor-1α (SDF-1α) is downregulated within diabetic cutaneous wounds, and that direct application of recombinant SDF-1α increases wound closure rates, neovascularization, and endothelial progenitor cell (EPC) recruitment. However, increased wound levels of exogenous SDF-1α results in elevated systemic levels of this proangiogenic chemokine that raises concerns for tumorigenesis and inflammation. We now seek to test the efficacy of a novel, safer cell-based therapy (CBT) employing ex vivo primed bone marrow-derived stem cells (BMDSC) with SDF-1α. We also elucidate the mechanism of action of this new approach for accelerating diabetic wound healing. METHODS: Unfractionated BMDSC from diabetic Lepr mice were incubated for 20 hours with SDF-1α (100 ng/mL) or bovine serum albumin (control). Pretreated BMDSC (1 × 10) were injected subcutaneously into full-thickness skin wounds in Lepr mice (n = 8 per group). Wound closure rates, capillary density, and the recruitment of EPC were assessed with serial photography, DiI perfusion, confocal microscopy, and immunohistochemistry. The expression of molecular targets, which may mediate prohealing/proangiogenic effects of SDF-1α-primed BMDSC was evaluated by polymerase chain reaction array and immunoblotting assay. The biological function of a potential mediator was tested in a mouse wound-healing model. Serum SDF-1α levels were measured with enzyme-linked immunosorbent assay (ELISA). RESULTS: SDF-1α-primed BMDSC significantly promote wound healing (P < 0.0001), neovascularization (P = 0.0028), and EPC recruitment (P = 0.0059). Gene/protein expression studies demonstrate upregulation of Ephrin Receptor B4 and plasminogen as downstream targets potentially mediating the prohealing and proangiogenic responses. Ex vivo BMDSC activation and the subsequent inoculation of cells into wounds does not increase systemic SDF-1α levels. CONCLUSIONS: We report a novel CBT that is highly effective in promoting healing and neovascularization in a murine model of type 2 diabetes. Furthermore, we identify new molecular targets that may be important for advancing the field of wound healing.


Assuntos
Transplante de Medula Óssea/métodos , Quimiocina CXCL12 , Diabetes Mellitus Tipo 2/complicações , Pele/lesões , Transplante de Células-Tronco/métodos , Cicatrização , Animais , Biomarcadores/sangue , Quimiocina CXCL12/sangue , Diabetes Mellitus Tipo 2/sangue , Modelos Animais de Doenças , Ensaio de Imunoadsorção Enzimática , Immunoblotting , Masculino , Camundongos , Microscopia Confocal , Neovascularização Fisiológica , Plasminogênio/metabolismo , Reação em Cadeia da Polimerase , Reepitelização , Pele/irrigação sanguínea , Fenômenos Fisiológicos da Pele , Lesões dos Tecidos Moles/terapia , Resultado do Tratamento , Cicatrização/fisiologia
17.
Ann Surg ; 254(3): 450-6; discussion 456-7, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21795970

RESUMO

OBJECTIVES: Aggressive human melanomas express, C-X-C chemokine receptor 4 (CXCR4), the receptor for the chemokine, stromal cell-derived factor-1alpha (SDF-1α). The CXCR4-SDF-1α axis has been postulated to increase melanoma invasiveness. We discovered that SDF-1α specifically upregulates E-selectin on endothelial cells, thus tethering circulating endothelial progenitor cells (EPC) and facilitating homing. We investigated the hypothesis that small interfering ribonucleic acid (siRNA)-mediated E-selectin blockade inhibits melanoma angiogenesis and tumor growth. METHODS: Human melanoma cells overexpressing SDF-1α were xenografted on severe combined immunodeficiency (SCID) mice. SDF-1α expression in cells was measured by enzyme-linked immunosorbent assay (ELISA). In vitro melanoma cell growth was examined by cell proliferation assay. In vivo vascular E-selectin knockdown was achieved by administration of high-volume E-selectin siRNA (100 pmol/180 µL/week × 3 times) and inhibition was validated by immunostaining (N = 6/group, E-Selectin siRNA vs control siRNA). Tumor angiogenesis was quantified (DiI-perfusion and LASER confocal microscopy). EPC homing to tumor vasculature was detected by immunostaining. Explanted in vivo tumor size and weight were measured. RESULTS: Three melanoma cells tested expressed undetectable levels of SDF-1α. Additional enforced overexpression of SDF-1α (by Lenti-SDF-1α) increased melanoma cell growth both in vitro and in vivo, enhanced EPC homing to tumor tissue, and increased tumor angiogenesis. Knocking-down vascular E-selectin significantly inhibited SDF-1α-induced EPC homing, tumor angiogenesis, and decreased melanoma growth in vivo. CONCLUSIONS: Downregulation of vascular E-selectin profoundly inhibits EPC homing, tumor angiogenesis, and tumor growth in human melanoma xenograft murine model, potentially by suppression of E-selectin-mediated EPC-endothelial cells interactions/homing. These findings identify E-selectin as a novel target for inhibition of melanoma angiogenesis and tumor growth.


Assuntos
Quimiocina CXCL12/efeitos dos fármacos , Selectina E/efeitos dos fármacos , Selectina E/genética , Melanoma Experimental/tratamento farmacológico , Melanoma Experimental/genética , Neovascularização Patológica/tratamento farmacológico , RNA Interferente Pequeno/uso terapêutico , Animais , Quimiocina CXCL12/metabolismo , Regulação para Baixo , Selectina E/metabolismo , Ensaio de Imunoadsorção Enzimática , Regulação Neoplásica da Expressão Gênica , Humanos , Melanoma Experimental/metabolismo , Camundongos , Camundongos SCID , Neovascularização Patológica/metabolismo , Transplante Heterólogo , Resultado do Tratamento , Regulação para Cima
18.
Ann Surg ; 252(4): 625-34, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20881769

RESUMO

OBJECTIVES: We previously reported that stromal cell-derived factor-1α (SDF-1α, a homing signal for recruiting endothelial progenitor cells (EPC) to areas of neovascularization), is down-regulated in diabetic wounds (Gallagher et al, J Clin Invest. 2007;117:1249-1259). We now investigate signals whereby mature endothelial cells (EC) and circulating EPC achieve SDF-1α-mediated EPC homing. METHODS: SDF-1α in diabetic wounds were therapeutically increased by injection of SDF-1α-engineered bone marrow-derived fibroblasts versus control cells (N = 48 [20, non-obese diabetic (NOD)], [28, streptozotocin-C57]). Polymerase chain reaction-array gene expression differences were validated by Western blotting and immunohistochemistry. The role of adhesion molecule(s) in mediating SDF-1α-induced EPC homing, and wound healing was furthered studied using antagonists in vitro and in vivo. RESULTS: Increasing wound SDF-1α via cell-based therapy promotes healing in diabetic mice (∼20% increase in healing rates by day 3, P = 0.006). SDF-1α increased EC-EPC adhesion and specifically upregulated E-selectin expression in human microvascular EC (2.3-fold increase, P < 0.01). This effect was also significant in blood vessels of the experimental mice and resulted in increased wound neovascularization. The regulatory effects of SDF-1α on EC-EPC adhesion and EPC homing were specifically mediated by E-selectin, as the application of E-selectin antagonists significantly inhibited SDF-1α-induced EC-EPC adhesion, EPC homing, wound neovascularization, and wound healing. CONCLUSIONS: SDF-1α-engineered cell-based therapy promotes diabetic wound healing in mice by specifically upregulating E-selectin expression in mature EC leading to increase EC-EPC adhesion, EPC homing, and increased wound neovascularization. These findings provide novel insight into the signals underlying the biological effect of SDF-1α on EPC homing and point to E-selectin as a new potential target for therapeutic manipulation of EPC trafficking in diabetic wound healing.


Assuntos
Diabetes Mellitus/fisiopatologia , Selectina E/fisiologia , Neovascularização Fisiológica/fisiologia , Cicatrização/fisiologia , Animais , Western Blotting , Adesão Celular/fisiologia , Células Cultivadas , Quimiocina CXCL12/fisiologia , Dependovirus , Diabetes Mellitus Experimental/fisiopatologia , Células Endoteliais/fisiologia , Feminino , Fibroblastos/fisiologia , Humanos , Imuno-Histoquímica , Fluxometria por Laser-Doppler , Lentivirus , Camundongos , Camundongos Endogâmicos C57BL , Microscopia Confocal , Reação em Cadeia da Polimerase , Células-Tronco/fisiologia
19.
Am J Physiol Renal Physiol ; 298(1): F125-32, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19864304

RESUMO

Glomerular endothelial cells (GEC) are strategically situated within the capillary loop and adjacent to the glomerular mesangium. GEC serve as targets of metabolic, biochemical, and hemodynamic signals that regulate the glomerular microcirculation. Unequivocally, hyperglycemia, hypertension, and the local renin-angiotensin system partake in the initiation and progression of diabetic nephropathy (DN). Whether free fatty acids (FFA) and reactive oxygen species (ROS) that have been associated with the endothelial dysfunction of diabetic macrovascular disease also contribute to DN is not known. Since endothelial cells from different organs and from different species may display different phenotypes, we employed human GEC to investigate the effect of high glucose (22.5 mmol/l), FFA (800 micromol/l), and angiotensin II (ANG II; 10(-7) mol/l) on the genesis of ROS and their effects on endothelial nitric oxide synthase (eNOS), cyclooxygenase-2 (COX-2), and the synthesis of prostaglandins (PGs). We demonstrated that high glucose but not high FFA increased the expression of a dysfunctional eNOS as well as increased ROS from NADPH oxidase (100%) and likely from uncoupled eNOS. ANG II also induced ROS from NADPH oxidase. High glucose and ANG II upregulated (100%) COX-2 via ROS and significantly increased the synthesis of prostacyclin (PGI(2)) by 300%. In contrast, FFA did not upregulate COX-2 but increased PGI(2) (500%). These novel studies are the first in human GEC that characterize the differential role of FFA, hyperglycemia, and ANG II on the genesis of ROS, COX-2, and PGs and their interplay in the early stages of hyperglcyemia.


Assuntos
Angiotensina II/fisiologia , Endotélio/metabolismo , Ácidos Graxos não Esterificados/fisiologia , Glomérulos Renais/metabolismo , Estresse Oxidativo/fisiologia , Angiotensina II/metabolismo , Células Cultivadas , Ciclo-Oxigenase 2/metabolismo , Relação Dose-Resposta a Droga , Endotélio/citologia , Epoprostenol/metabolismo , Glucose/fisiologia , Humanos , Hiperglicemia/metabolismo , Glomérulos Renais/citologia , NADPH Oxidases/metabolismo , Óxido Nítrico Sintase Tipo III/metabolismo , Oxigênio/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Superóxidos/metabolismo
20.
Am J Nephrol ; 29(4): 319-26, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-18849602

RESUMO

BACKGROUND/AIMS: Epidemiologic studies suggest that cigarette smoke worsens the progression of renal injury in patients with glomerular diseases. The mechanisms involved have not been elucidated. These studies were designed to determine whether nicotine worsens markers of inflammation including glomerular cell proliferation and fibronectin deposition in an in vivo model of glomerular injury. METHODS: Sprague-Dawley rats were injected with anti-Thy1 antibody and given either tap water or nicotine in the drinking water until sacrifice at day 14. Fibronectin expression was measured by Western blot and immunohistochemistry. COX-2 expression was also determined by Western blot in the kidney cortex of rats treated with nicotine and in cultured human mesangial cells treated with nicotine. RESULTS: Anti-Thy1 antibody administration resulted in a significant increase in the number of cells per glomerulus that was further increased by the administration of nicotine. In nephritic rats, the administration of nicotine significantly increased fibronectin and COX-2 expression. In cultured human mesangial cells we also demonstrated that nicotine increases COX-2 expression and activity and that COX-2 mediates mesangial cell proliferation in response to nicotine. CONCLUSION: Either in vivo or in vitro treatment with nicotine leads to activation of inflammatory mediators and hallmarks of glomerular injury, which may explain the mechanisms involved in the deleterious effects of cigarette smoking on renal disease.


Assuntos
Glomerulonefrite/patologia , Células Mesangiais/efeitos dos fármacos , Células Mesangiais/patologia , Nicotina/toxicidade , Agonistas Nicotínicos/toxicidade , Doença Aguda , Animais , Contagem de Células , Divisão Celular/efeitos dos fármacos , Ciclo-Oxigenase 2/metabolismo , Modelos Animais de Doenças , Progressão da Doença , Fibronectinas/metabolismo , Glomerulonefrite/metabolismo , Isoanticorpos/farmacologia , Células Mesangiais/metabolismo , Prostaglandinas/metabolismo , Ratos , Ratos Sprague-Dawley , Fumar/efeitos adversos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA